Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Microbiol ; 15: 1367898, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38511003

RESUMEN

The gut represents an important site of colonization of the commensal bacterium Streptococcus agalactiae (group B Streptococcus or GBS), which can also behave as a deadly pathogen in neonates and adults. Invasion of the intestinal epithelial barrier is likely a crucial step in the pathogenesis of neonatal infections caused by GBS belonging to clonal complex 17 (CC17). We have previously shown that the prototypical CC17 BM110 strain invades polarized enterocyte-like cells through their lateral surfaces using an endocytic pathway. By analyzing the cellular distribution of putative GBS receptors in human enterocyte-like Caco-2 cells, we find here that the alpha 3 (α3) and alpha 2 (α2) integrin subunits are selectively expressed on lateral enterocyte surfaces at equatorial and parabasal levels along the vertical axis of polarized cells, in an area corresponding to GBS entry sites. The α3ß1 and α2ß1 integrins were not readily accessible in fully differentiated Caco-2 monolayers but could be exposed to specific antibodies after weakening of intercellular junctions in calcium-free media. Under these conditions, anti-α3ß1 and anti-α2ß1 antibodies significantly reduced GBS adhesion to and invasion of enterocytes. After endocytosis, α3ß1 and α2ß1 integrins localized to areas of actin remodeling around GBS containing vacuoles. Taken together, these data indicate that GBS can invade enterocytes by binding to α3ß1 and α2ß1 integrins on the lateral membrane of polarized enterocytes, resulting in cytoskeletal remodeling and bacterial internalization. Blocking integrins might represent a viable strategy to prevent GBS invasion of gut epithelial tissues.

2.
Cell Rep Med ; 4(7): 101098, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37390829

RESUMEN

During differentiation, neutrophils undergo a spontaneous pro-inflammatory program that is hypothesized here to be under caspase-8 control. In mice, intraperitoneal administration of the caspase-8 inhibitor z-IETD-fmk is sufficient to unleash the production of pro-inflammatory cytokines and neutrophil influx in the absence of cell death. These effects are due to selective inhibition of caspase-8 and require tonic interferon-ß (IFN-ß) production and RIPK3 but not MLKL, the essential downstream executioner of necroptotic cell death. In vitro, stimulation with z-IETD-fmk is sufficient to induce significant cytokine production in murine neutrophils but not in macrophages. Therapeutic administration of z-IETD-fmk improves clinical outcome in models of lethal bacterial peritonitis and pneumonia by augmenting cytokine release, neutrophil influx, and bacterial clearance. Moreover, the inhibitor protects mice against high-dose endotoxin shock. Collectively, our data unveil a RIPK3- and IFN-ß-dependent pathway that is constitutively activated in neutrophils and can be harnessed therapeutically using caspase-8 inhibition.


Asunto(s)
Apoptosis , Infecciones Bacterianas , Animales , Ratones , Infecciones Bacterianas/tratamiento farmacológico , Caspasa 8/metabolismo , Caspasa 8/farmacología , Citocinas/metabolismo , Activación Neutrófila
3.
Antibiotics (Basel) ; 12(6)2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37370284

RESUMEN

The number of multidrug-resistant bacteria is rapidly spreading worldwide. Among the various mechanisms determining resistance to antimicrobial agents, multidrug efflux pumps play a noteworthy role because they export extraneous and noxious substrates from the inside to the outside environment of the bacterial cell contributing to multidrug resistance (MDR) and, consequently, to the failure of anti-infective therapies. The expression of multidrug efflux pumps can be under the control of transcriptional regulators and two-component systems (TCS). TCS are a major mechanism by which microorganisms sense and reply to external and/or intramembrane stimuli by coordinating the expression of genes involved not only in pathogenic pathways but also in antibiotic resistance. In this review, we describe the influence of TCS on multidrug efflux pump expression and activity in some Gram-negative and Gram-positive bacteria. Taking into account the strict correlation between TCS and multidrug efflux pumps, the development of drugs targeting TCS, alone or together with already discovered efflux pump inhibitors, may represent a beneficial strategy to contribute to the fight against growing antibiotic resistance.

4.
Pathogens ; 12(1)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36678467

RESUMEN

Two-component signaling systems (TCSs) are finely regulated mechanisms by which bacteria adapt to environmental conditions by modifying the expression of target genes. In bacterial pathogenesis, TCSs play important roles in modulating adhesion to mucosal surfaces, resistance to antibiotics, and metabolic adaptation. In the context of urinary tract infections (UTI), one of the most common types infections causing significant health problems worldwide, uropathogens use TCSs for adaptation, survival, and establishment of pathogenicity. For example, uropathogens can exploit TCSs to survive inside bladder epithelial cells, sense osmolar variations in urine, promote their ascension along the urinary tract or even produce lytic enzymes resulting in exfoliation of the urothelium. Despite the usefulness of studying the function of TCSs in in vitro experimental models, it is of primary necessity to study bacterial gene regulation also in the context of host niches, each displaying its own biological, chemical, and physical features. In light of this, the aim of this review is to provide a concise description of several bacterial TCSs, whose activity has been described in mouse models of UTI.

5.
Mol Microbiol ; 117(6): 1479-1492, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35570359

RESUMEN

The binding of Streptococcus pneumoniae to collagen is likely an important step in the pathogenesis of pneumococcal infections, but little is known of the underlying molecular mechanisms. Streptococcal surface repeats (SSURE) are highly conserved protein domains present in cell wall adhesins from different Streptococcus species. We find here that SSURE repeats of the pneumococcal adhesin plasminogen and fibronectin binding protein B (PfbB) bind to various types of collagen. Moreover, deletion of the pfbB gene resulted in a significant impairment of the ability of encapsulated or unencapsulated pneumococci to bind collagen. Notably, a PfbB SSURE domain is also bound to the complement component C1q that bears a collagen-like domain and promotes adherence of pneumococci to host cells by acting as a bridge between bacteria and epithelial cells. Accordingly, deletion of PfbB or pre-treatment with anti-SSURE antibodies markedly decreased pneumococcal binding to C1q as well as C1q-dependent adherence to epithelial and endothelial cells. Further data indicated that C1q promotes pneumococcal adherence by binding to integrin α2 ß1 . In conclusion, our results indicate that the SSURE domains of the PfbB protein promote interactions of pneumococci with various types of collagen and with C1q. These repeats may be useful targets in strategies to control S. pneumoniae infections.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Colágeno/genética , Colágeno/metabolismo , Complemento C1q/genética , Complemento C1q/metabolismo , Células Endoteliales/metabolismo , Humanos , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo
6.
Front Microbiol ; 13: 881549, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572655

RESUMEN

Group B Streptococcus (GBS) is a Gram-positive bacterium able to switch from a harmless commensal of healthy adults to a pathogen responsible for invasive infections in neonates. The signals and regulatory mechanisms governing this transition are still largely unknown. CodY is a highly conserved global transcriptional regulator that links nutrient availability to the regulation of major metabolic and virulence pathways in low-G+C Gram-positive bacteria. In this work, we investigated the role of CodY in BM110, a GBS strain representative of a hypervirulent lineage associated with the majority of neonatal meningitis. Deletion of codY resulted in a reduced ability of the mutant strain to cause infections in neonatal and adult animal models. The observed decreased in vivo lethality was associated with an impaired ability of the mutant to persist in the blood, spread to distant organs, and cross the blood-brain barrier. Notably, the codY null mutant showed reduced adhesion to monolayers of human epithelial cells in vitro and an increased ability to form biofilms, a phenotype associated with strains able to asymptomatically colonize the host. RNA-seq analysis showed that CodY controls about 13% of the genome of GBS, acting mainly as a repressor of genes involved in amino acid transport and metabolism and encoding surface anchored proteins, including the virulence factor Srr2. CodY activity was shown to be dependent on the availability of branched-chain amino acids, which are the universal cofactors of this regulator. These results highlight a key role for CodY in the control of GBS virulence.

7.
EMBO J ; 41(5): e109386, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35112724

RESUMEN

The mechanisms whereby neutrophils respond differentially to live and dead organisms are unknown. We show here that neutrophils produce 5- to 30-fold higher levels of the Cxcl2 chemokine in response to live bacteria, compared with killed bacteria or isolated bacterial components, despite producing similar levels of Cxcl1 or pro-inflammatory cytokines. Secretion of high levels of Cxcl2, which potently activates neutrophils by an autocrine mechanism, requires three signals. The first two signals are provided by two different sets of signal peptides released by live bacteria, which selectively activate formylated peptide receptor 1 (Fpr1) and Fpr2, respectively. Signal 3 originates from Toll-like receptor activation by microbial components present in both live and killed bacteria. Mechanistically, these signaling pathways converge at the level of the p38 MAP kinase, leading to activation of the AP-1 transcription factor and to Cxcl2 induction. Collectively, our data demonstrate that the simultaneous presence of agonists for Fpr1, Fpr2, and Toll-like receptors represents a unique signature associated with viable bacteria, which is sensed by neutrophils and induces Cxcl2-dependent autocrine cell activation.


Asunto(s)
Bacterias/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Receptores Toll-Like/metabolismo , Animales , Citocinas/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
8.
Front Cell Infect Microbiol ; 11: 679792, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34568085

RESUMEN

Binding to plasminogen (Plg) enables bacteria to associate with and invade host tissues. The cell wall protein PbsP significantly contributes to the ability of group B streptococci, a frequent cause of invasive infection, to bind Plg. Here we sought to identify the molecular regions involved in the interactions between Plg and PbsP. The K4 Kringle domain of the Plg molecule was required for binding of Plg to whole PbsP and to a PbsP fragment encompassing a region rich in methionine and lysine (MK-rich domain). These interactions were inhibited by free L-lysine, indicating the involvement of lysine binding sites in the Plg molecule. However, mutation to alanine of all lysine residues in the MK-rich domain did not decrease its ability to bind Plg. Collectively, our data identify a novel bacterial sequence that can interact with lysine binding sites in the Plg molecule. Notably, such binding did not require the presence of lysine or other positively charged amino acids in the bacterial receptor. These data may be useful for developing alternative therapeutic strategies aimed at blocking interactions between group B streptococci and Plg.


Asunto(s)
Lisina , Plasminógeno , Sitios de Unión , Pared Celular/metabolismo , Lisina/metabolismo , Plasminógeno/metabolismo , Unión Proteica , Streptococcus agalactiae
9.
J Immunol ; 207(5): 1448-1455, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34362834

RESUMEN

Identification of the receptors involved in innate immune recognition of Staphylococcus aureus, a major cause of morbidity and mortality in humans, is essential to develop alternative strategies to treat infections caused by antibiotic-resistant strains. In the current study, we examine the role of endosomal TLRs, which sense the presence of prokaryotic-type nucleic acids, in anti-staphylococcal host defenses using infection models involving genetically defective mice. Single deficiencies in TLR7, 9, or 13 resulted in mild or no decrease in host defenses. However, the simultaneous absence of TLR7, 9, and 13 resulted in markedly increased susceptibility to cutaneous and systemic S. aureus infection concomitantly with decreased production of proinflammatory chemokines and cytokines, neutrophil recruitment to infection sites, and reduced production of reactive oxygen species. This phenotype was significantly more severe than that of mice lacking TLR2, which senses the presence of staphylococcal lipoproteins. Notably, the combined absence of TLR7, 9, and 13 resulted in complete abrogation of IL-12 p70 and IFN-ß responses to staphylococcal stimulation in macrophages. Taken together, our data highlight the presence of a highly integrated endosomal detection system, whereby TLR7, 9, and 13 cooperate in sensing the presence of staphylococcal nucleic acids. We demonstrate that the combined absence of these receptors cannot be compensated for by cell surface-associated TLRs, such as TLR2, or cytosolic receptors. These data may be useful to devise strategies aimed at stimulating innate immune receptors to treat S. aureus infections.


Asunto(s)
Endosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética , Receptores Toll-Like/genética
10.
PLoS One ; 16(6): e0253242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34129624

RESUMEN

Streptococcus agalactiae (group B streptococcus or GBS) is a commensal bacterium that can frequently behave as a pathogen, particularly in the neonatal period and in the elderly. The gut is a primary site of GBS colonization and a potential port of entry during neonatal infections caused by hypervirulent clonal complex 17 (CC17) strains. Here we studied the interactions between the prototypical CC17 BM110 strain and polarized enterocytes using the Caco-2 cell line. GBS could adhere to and invade these cells through their apical or basolateral surfaces. Basolateral invasion was considerably more efficient than apical invasion and predominated under conditions resulting in weakening of cell-to-cell junctions. Bacterial internalization occurred by a mechanism involving caveolae- and lipid raft-dependent endocytosis and actin re-organization, but not clathrin-dependent endocytosis. In the first steps of Caco-2 invasion, GBS colocalized with the early endocytic marker EEA-1, to later reside in acidic vacuoles. Taken together, these data suggest that CC17 GBS selectively adheres to the lateral surface of enterocytes from which it enters through caveolar lipid rafts using a classical, actin-dependent endocytic pathway. These data may be useful to develop alternative preventive strategies aimed at blocking GBS invasion of the intestinal barrier.


Asunto(s)
Enterocitos/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/patogenicidad , Adhesión Bacteriana , Células CACO-2/microbiología , Endocitosis , Humanos , Uniones Intercelulares/microbiología , Microscopía Fluorescente , Streptococcus agalactiae/fisiología , Virulencia
11.
Pathogens ; 10(2)2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525468

RESUMEN

Previous studies performed using a model of group B streptococcus (GBS)-induced peritoneal inflammation indicate that the interleukin-1 receptor (IL-1R) family plays an important role in the innate host defense against this encapsulated Gram-positive bacteria. Since the role of IL-1-dependent signaling in peritoneal infections induced by other Gram-positive bacteria is unknown, in the present study we sought to investigate the contribution of IL-1R signaling in host defenses against Streptococcus pyogenes (group A streptococcus or GAS) or Staphylococcus aureus, two frequent and global human Gram-positive extracellular pathogens. We analyzed here the outcome of GAS or S. aureus infection in IL-1R-deficient mice. After inoculated intraperitoneal (i.p.) inoculation with group A Streptococcus or S. aureus, all the wild-type (WT) control mice survived the challenge, while, respectively, 63% or 50% of IL-1-defective mice died. Lethality was due to the ability of both bacterial species to replicate and disseminate to the target organs of IL-1R-deficient mice. Moreover, the experimental results indicate that IL-1 signaling promotes the production of leukocyte attractant chemokines CXCL-1 and CXCL-2 and recruitment of neutrophils to bacterial infection sites. Accordingly, the reduced neutrophil recruitment in IL-1R-deficient mice was linked with decreased production of neutrophil chemokines. Collectively, our findings indicate that IL-1 signaling, as previously showed in host defense against GBS, plays a fundamental role also in controlling the progression and outcome of GAS or S. aureus disease.

12.
Med Mycol ; 58(8): 1138-1148, 2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-32246714

RESUMEN

Members of the C. neoformans/C. gattiii species complex are an important cause of serious humans infections, including meningoencephalitis. We describe here a 45 kDa extracellular cellulase purified from culture supernatants of C. neoformans var. neoformans. The N-terminal sequence obtained from the purified protein was used to isolate a clone containing the full-length coding sequence from a C. neoformans var. neoformans (strain B-3501A) cDNA library. Bioinformatics analysis indicated that this gene is present, with variable homology, in all sequenced genomes of the C. neoformans/C. gattii species complex. The cDNA clone was used to produce a recombinant 45 kDa protein in E. coli that displayed the ability to convert carboxymethyl cellulose and was therefore designated as NG-Case (standing for Neoformans Gattii Cellulase). To explore its potential use as a vaccine candidate, the recombinant protein was used to immunize mice and was found capable of inducing T helper type 1 responses and delayed-type hypersensitivity reactions, but not immune protection against a highly virulent C. neoformans var grubii strain. These data may be useful to better understand the mechanisms underlying the ability C. neoformans/C. gattii to colonize plant habitats and to interact with the human host during infection.


Asunto(s)
Celulasa/inmunología , Cryptococcus/enzimología , Proteínas Fúngicas/inmunología , Animales , Carboximetilcelulosa de Sodio/metabolismo , Celulasa/química , Celulasa/genética , Celulasa/metabolismo , Criptococosis/inmunología , Criptococosis/microbiología , Cryptococcus/genética , Cryptococcus/inmunología , Cryptococcus/metabolismo , Cryptococcus neoformans/enzimología , Cryptococcus neoformans/genética , Cryptococcus neoformans/inmunología , Cryptococcus neoformans/metabolismo , Medios de Cultivo Condicionados , Citocinas/inmunología , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Inmunización , Ratones , Peso Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Células TH1/inmunología
13.
mBio ; 11(2)2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209688

RESUMEN

Streptococcus pneumoniae (or pneumococcus) is a highly prevalent human pathogen. Toll-like receptors (TLRs) function as immune sensors that can trigger host defenses against this bacterium. Defects in TLR-activated signaling pathways, including deficiency in the adaptor protein myeloid differentiation factor 88 (MyD88), are associated with markedly increased susceptibility to infection. However, the individual MyD88-dependent TLRs predominantly involved in antipneumococcal defenses have not been identified yet. Here we find that triple knockout mice simultaneously lacking TLR7, TLR9, and TLR13, which sense the presence of bacterial DNA (TLR9) and RNA (TLR7 and TLR13) in the phagolysosomes of phagocytic cells, display a phenotype that largely resembles that of MyD88-deficient mice and rapidly succumb to pneumococcal pneumonitis due to defective neutrophil influx into the lung. Accordingly, TLR7/9/13 triple knockout resident alveolar macrophages were largely unable to respond to pneumococci with the production of neutrophil-attracting chemokines and cytokines. Mice with single deficiencies of TLR7, TLR9, or TLR13 showed unaltered ability to control lung infection but were moderately more susceptible to encephalitis, in association with a decreased ability of microglia to mount cytokine responses in vitro Our data point to a dominant, tissue-specific role of nucleic acid-sensing pathways in innate immune recognition of S. pneumoniae and also show that endosomal TLRs are largely capable of compensating for the absence of each other, which seems crucial to prevent pneumococci from escaping immune recognition. These results may be useful to develop novel strategies to treat infections by antibiotic-resistant pneumococci based on stimulation of the innate immune system.IMPORTANCE The pneumococcus is a bacterium that frequently causes infections in the lungs, ears, sinus cavities, and meninges. During these infections, body defenses are triggered by tissue-resident cells that use specialized receptors, such as Toll-like receptors (TLRs), to sense the presence of bacteria. We show here that pneumococci are predominantly detected by TLRs that are located inside intracellular vacuoles, including endosomes, where these receptors can sense the presence of nucleic acids released from ingested bacteria. Mice that simultaneously lacked three of these receptors (specifically, TLR7, TLR9, and TLR13) were extremely susceptible to lung infection and rapidly died after inhalation of pneumococci. Moreover, tissue-resident macrophages from these mice were impaired in their ability to respond to the presence of pneumococci by producing inflammatory mediators capable of recruiting polymorphonuclear leucocytes to infection sites. This information may be useful to develop drugs to treat pneumococcal infections, particularly those caused by antibiotic-resistant strains.


Asunto(s)
Inmunidad Innata , Ácidos Nucleicos/inmunología , Streptococcus pneumoniae/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Animales , Citocinas/inmunología , Femenino , Pulmón/inmunología , Pulmón/microbiología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Infiltración Neutrófila , Transducción de Señal , Streptococcus pneumoniae/patogenicidad , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética
14.
J Immunol ; 204(3): 660-670, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31852751

RESUMEN

The influx of neutrophils to infection sites is a fundamental step in host defenses against the frequent human pathogen group B Streptococcus (GBS) and other extracellular bacteria. Using a mouse model of GBS-induced peritonitis, we show in this study that the chemokines Cxcl1 and Cxcl2 play distinctive roles in enhancing the recruitment and the antibacterial activities of neutrophils in a manner that is linked to differences in the cellular sources of these mediators. Cell depletion experiments demonstrated that neutrophils make a significant contribution to the in vivo production of Cxcl2 but not Cxcl1. In vitro, neutrophils responded weakly to LPS but released high levels of Cxcl2 after stimulation with GBS or other bacteria. Neutrophil-derived Cxcl2 acted in an autocrinous manner to increase its own production and to enhance antibacterial activities, including the release of oxygen radicals. In both neutrophils and macrophages, the production of Cxcl1/2 largely required the presence of functional UNC93B1, a chaperone protein involved in signaling by endosomal TLRs. Moreover, the phenotype of UNC93B1-defective phagocytes could be recapitulated by the simultaneous absence of TLR7, 9, and 13 but not by the absence of individual TLRs. Collectively, our data show that neutrophils recognize Gram-positive and Gram-negative bacteria by means of multiple phagosomal TLRs, resulting in de novo synthesis of Cxcl2, amplification of neutrophil recruitment, and potentiation of their antibacterial activities. These data may be useful to devise alternative therapeutic strategies aimed at enhancing the recruitment and the functional activities of polymorphonuclear leukocytes during infections caused by antibiotic-resistant bacteria.


Asunto(s)
Infecciones Bacterianas/inmunología , Quimiocina CXCL2/metabolismo , Endosomas/metabolismo , Neutrófilos/inmunología , Peritonitis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Receptores Toll-Like/metabolismo
15.
Trends Microbiol ; 27(9): 727-728, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31324435

RESUMEN

Streptococcus agalactiae meningitis is a frequent neonatal disease associated with high mortality and permanent neurological damage. Deng et al. (PLoS Pathog., 2019) now show that interactions between the bacterial protein BspC and host cell vimentin participate in the process of invasion of the meninges by this bacterial pathogen.


Asunto(s)
Meningitis , Infecciones Estreptocócicas , Antígenos de Superficie , Encéfalo , Endotelio , Humanos , Inflamación , Streptococcus agalactiae , Vimentina
16.
Biomed J ; 42(1): 8-18, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30987709

RESUMEN

One of the distinguishing features of the 1918 pandemic is the occurrence of massive, potentially detrimental, activation of the innate immune system in critically ill patients. Whether this reflects an intrinsic capacity of the virus to induce an exaggerated inflammatory responses or its remarkable ability to reproduce in vivo is still open to debate. Tremendous progress has recently been made in our understanding of innate immune responses to influenza infection and it is now time to translate this knowledge into therapeutic strategies, particularly in view of the possible occurrence of future outbreaks caused by virulent strains.


Asunto(s)
Brotes de Enfermedades/historia , Inmunidad Innata/inmunología , Gripe Humana/inmunología , Gripe Humana/terapia , Animales , Historia del Siglo XX , Humanos , Inflamasomas/inmunología , Gripe Humana/historia , Transducción de Señal/inmunología
17.
Sci Rep ; 8(1): 14322, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254272

RESUMEN

Streptococcus agalactiae (Group B Streptococcus or GBS) is a frequent cause of serious disease in newborns and adults. Epidemiological evidence indicates a strong association between GBS strains belonging to the hypervirulent CC17 clonal complex and the occurrence of meningitis in neonates. We investigate here the role of PbsP, a cell wall plasminogen binding protein, in colonization of the central nervous system by CC17 GBS. Deletion of pbsP selectively impaired the ability of the CC17 strain BM110 to colonize the mouse brain after intravenous challenge, despite its unchanged capacity to persist at high levels in the blood and to invade the kidneys. Moreover, immunization with a recombinant form of PbsP considerably reduced brain infection and lethality. In vitro, pbsP deletion markedly decreased plasmin-dependent transmigration of BM110 through brain microvascular endothelial cells. Although PbsP was modestly expressed in bacteria grown under standard laboratory conditions, pbsP expression was markedly upregulated during in vivo infection or upon contact with cultured brain endothelial cells. Collectively, our studies indicate that PbsP is a highly conserved Plg binding adhesin, which is functionally important for invasion of the central nervous system by the hypervirulent CC17 GBS. Moreover, this antigen is a promising candidate for inclusion in a universal GBS vaccine.


Asunto(s)
Proteínas Bacterianas/metabolismo , Encéfalo/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Animales , Encéfalo/citología , Movimiento Celular , Células Endoteliales/citología , Fibrinolisina/metabolismo , Regulación Bacteriana de la Expresión Génica , Ratones , Streptococcus agalactiae/genética , Virulencia
18.
Intervirology ; 61(1): 1-8, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30021203

RESUMEN

BACKGROUND: Hepatitis C virus (HCV) NS3 resistance-associated substitutions (RASs) reduce HCV susceptibility to protease inhibitors. Little is known about NS3 RASs in viral isolates from the liver of chronic hepatitis C (CHC) patients infected with HCV genotype-1a (G1a). AIM: The objective of this work was to study NS3 variability in isolates from the serum and liver of HCV-G1a-infected patients naïve to direct-acting antivirals (DAAs). METHODS: NS3 variability of HCV-G1a isolates from the serum and liver of 11 naïve CHC patients, and from sera of an additional 20 naïve CHC patients, was investigated by next-generation sequencing. RESULTS: At a cutoff of 1%, NS3 RASs were detected in all the samples examined. At a cutoff of 15%, they were found in 54.5% (6/11) and 27.3% (3/11) of the paired liver and serum samples, respectively, and in 22.5% (7/31) of the overall serum samples examined. Twenty-six out of thirty-one (84%) patients showed NS3 variants with multiple RASs. Phylogenetic analysis showed that NS3 sequences clustered within 2 clades, with 10/31 (32.2%) patients infected by clade I, 15/31 (48.8%) by clade II, and 6/31 (19.3%) by both clades. CONCLUSIONS: Though the number of patients examined was limited, NS3 variants with RASs appear to be major components of both intrahepatic and circulating viral quasispecies populations in DAA-naïve patients.


Asunto(s)
Variación Genética , Hepacivirus/enzimología , Hepatitis C Crónica/virología , Proteínas no Estructurales Virales/genética , Adulto , Sustitución de Aminoácidos , Antivirales/farmacología , Farmacorresistencia Viral , Femenino , Genotipo , Hepacivirus/genética , Hepatitis C Crónica/epidemiología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Italia/epidemiología , Hígado/virología , Masculino , Persona de Mediana Edad , Filogenia , Inhibidores de Proteasas/farmacología , Suero/virología
19.
Mol Microbiol ; 110(1): 82-94, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30030946

RESUMEN

Binding of microbial pathogens to host vitronectin (Vtn) is a common theme in the pathogenesis of invasive infections. In this study, we characterized the role of Vtn in the invasion of mucosal epithelial cells by Streptococcus agalactiae (i.e. group B streptococcus or GBS), a frequent human pathogen. Moreover, we identified PbsP, a previously described plasminogen-binding protein of GBS, as a dual adhesin that can also interact with human Vtn through its streptococcal surface repeat (SSURE) domains. Deletion of the pbsP gene decreases both bacterial adhesion to Vtn-coated inert surfaces and the ability of GBS to interact with epithelial cells. Bacterial adherence to and invasion of epithelial cells were either inhibited or enhanced by cell pretreatment with, respectively, anti-Vtn antibodies or Vtn, confirming the role of Vtn as a GBS ligand on host cells. Finally, antibodies directed against the integrin αv subunit inhibited Vtn-dependent cell invasion by GBS. Collectively, these results indicate that Vtn acts as a bridge between the SSURE domains of PbsP on the GBS surface and host integrins to promote bacterial invasion of epithelial cells. Therefore, inhibition of interactions between PbsP and extracellular matrix components could represent a viable strategy to prevent colonization and invasive disease by GBS.


Asunto(s)
Proteínas Bacterianas/metabolismo , Integrina alfaV/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Vitronectina/metabolismo , Células A549 , Adhesión Bacteriana/genética , Proteínas Bacterianas/genética , Células CACO-2 , Pared Celular/metabolismo , Células Epiteliales/microbiología , Humanos , Integrina alfaV/genética , Dominios Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptococcus agalactiae/genética , Vitronectina/genética
20.
Int Immunopharmacol ; 62: 86-95, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29990698

RESUMEN

Genistein, a natural isoflavone found in soybean products, is considered as a powerful anti-cancer agent, although the involved mechanisms are not fully understood. There is a growing body of evidence that, among the genes inhibited by genistein and responsible for cell cycle progression, invasion, metastasis, and angiogenesis, IL-8 occupies a relevant place. On the other hand, it is equally well documented that IL-8 is upregulated by prostaglandin E2 (PGE2) in different pathological conditions, particularly in neoplastic disease. Here we investigated whether genistein could affect cell growth in a panel of oral, uveal and cutaneous melanoma cell lines by interfering with basal or PGE2-induced IL-8 production. To this end, experiments were performed to evaluate the effect of PGE2 treatment on IL-8 levels, the expression and the role of PGE2 receptors and whether genistein could be able to interfere with these events. Finally, it was evaluated whether the inhibition of oral, uveal and cutaneous melanoma cell proliferation in the presence of genistein could be related to a reduction of IL-8 levels. We show that PGE2 enhances IL-8 synthesis via the EP3 receptor and that genistein is able to down-regulate the latter, as well as to decrease IL-8 mRNA and protein expression, thereby inhibiting oral, uveal and cutaneous melanoma cell proliferation. Taken together, our data provide new insights into the anti-cancer properties of genistein by showing that this flavonoid may affect the development and growth of melanoma at oral, uveal and cutaneous sites. Moreover, these results provide evidence that genistein may exert its therapeutic activity through its ability to prevent PGE2-mediated IL-8 induction.


Asunto(s)
Anticarcinógenos/farmacología , Antineoplásicos Fitogénicos/farmacología , Proliferación Celular/efectos de los fármacos , Dinoprostona/metabolismo , Genisteína/farmacología , Interleucina-8/antagonistas & inhibidores , Melanoma , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Dinoprostona/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...